T Cell Mediated Immunity to Influenza

download T Cell Mediated Immunity to Influenza

of 7

Transcript of T Cell Mediated Immunity to Influenza

  • 8/11/2019 T Cell Mediated Immunity to Influenza

    1/7

    T cell mediated immunity to influenza:mechanisms of viral control

    Nicole L. La Gruta and Stephen J. TurnerDepartment of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, The University of

    Melbourne, Parkville, Victoria, 3010, Australia

    Infection with influenza A virus (IAV) is a major cause

    of worldwide morbidity and mortality. Recent findings

    indicate

    that

    T

    cell immunity is

    key to

    limiting

    severity

    of disease arising from IAV infection, particularly in

    instances where antibody immunity is ineffective. As

    such, there is

    a

    need to

    understand

    better

    the

    mecha-

    nisms that mediate effective IAV-specific cellular immu-

    nity, especially given that T cell immunity must form an

    integral part of any vaccine designed to elicit crossreac-tive immunity against existing and new strains of influ-

    enza virus. Here, we review the current understanding

    of cellular immunity to IAV, highlighting recent findings

    that demonstrate important roles for bothCD4+ and CD8+

    T cell immunity in protection from IAV-mediated disease.

    T cells

    and

    immunity

    to

    IAV

    infection

    Worldwide,

    seasonal

    IAV

    infection

    is

    a

    major

    cause

    of

    morbidity and mortality, estimated to be responsible for

    35

    million

    cases

    of

    severe

    illness

    and 250 000500 000

    deaths

    worldwide

    per

    annum

    (WHO

    influenza

    centre

    web-

    site). IAV-specific immunity can be induced byvaccination

    that

    generates

    IAV-specific

    antibodies

    that

    limit

    or

    preventIAV infection. However, the IAVvaccine needs to be refor-

    mulated

    on

    an

    annual

    basis

    because

    influenza

    viruses

    rapidly

    evolve,

    with

    new

    strains

    emerging

    that

    have

    lost

    or mutated the targets recognized by the preceding anti-

    body

    response.

    Thus,

    an

    arms

    race

    ensues

    whereby

    vac-

    cine-induced immune pressures select for new strains that

    are

    no

    longer

    recognized

    by

    vaccine

    induced

    IAV-specific

    antibodies,

    necessitating

    the

    production

    of

    updated

    IAV

    vaccines. CD8+ and CD4+ T cells have distinct but impor-

    tant

    roles

    in

    the

    control,

    and

    eventual

    clearance,

    of

    influ-

    enza virus infection [1]. Upon activation (Box 1), CD4+ T

    cells (or helper T cells) are thought to promote effective

    immunity

    primarily

    by

    providing

    the

    necessary

    secondary

    signals for optimal antibody responses, as well as produc-

    ing

    antiviral

    and

    proinflammatory

    cytokines

    upon

    infec-

    tion, although recent data indicate their role may extend

    beyond

    just

    cytokine

    production

    [2]. CD8+ T

    cells

    are

    often

    considered

    the

    hit-men

    of

    the

    immune

    system

    because

    they locate and kill virus-infected cells in the body, thus

    limiting

    viral

    spread

    and

    contributing

    to

    the

    eventual

    clearance

    of

    infection.

    CD8+ T

    cells

    express

    a

    range

    of

    effector genes including granzymes and perforin, which

    mediate

    their

    signature

    cytotoxic

    capacity.

    Given that processing

    and presentation

    of

    viral

    peptide

    targetson thehostcell surface can only occurafter infection,

    unlike

    preformed

    antibody responses,

    pre-existing

    T

    cell

    immunity

    cannot prevent

    IAV

    infectionper se; an issue that

    has, in the past, resulted in the dismissal of cellular immu-

    nity as a goal of effective vaccination.However, theutility ofcellular immunity

    stems

    from

    the fact that unlike antibody

    responses, cellular immunity targets viral proteins that are

    more likely to be shared betweendifferentvirus strains and

    subtypes [1,3], thereby offering a greater breadth of protec-

    tion. Moreover, unlike for

    chronic

    viruses

    such

    as

    HIV or

    hepatitis B virus, wherea primary goal ofvaccination must

    be

    sterilizing immunity,

    for

    acute viruses

    such

    as

    IAV,

    the

    principal

    objective

    is

    the amelioration of

    infection-associat-

    ed pathology until virus is cleared. Thus, it is widely ac-

    knowledged

    thata

    comprehensive

    vaccineagainst

    IAV

    must

    include

    the ability to

    elicit T

    cell

    immunity

    [4].

    Here, we

    examine the current state of knowledge regarding IAV-

    specific

    T

    cell immunity

    and discuss

    how

    a

    greater under-standing of factors that shape and promote IAV-specific

    cellular immunity

    will

    contribute

    to

    improved

    vaccinestrat-

    egies capable of eliciting heterologous immunity.

    Targets of the T cell response during influenza infection

    The factthatIAV-specific memory T cells cantarget a broad

    range of

    peptides

    derived

    from

    proteins

    that are relatively

    conserved betweendifferent

    influenzastrains and

    subtypes

    means that T cell immunity induced by one IAV strain has

    the

    potential

    to

    provide immunity

    against

    distinct IAV

    strains

    in

    the absence

    of

    neutralizing antibody

    (termed

    heterologous

    immunity).

    If

    we

    are to

    take full

    advantage

    of

    IAV-specific

    T

    cell immunity

    via

    development

    of a

    novelT

    cellbasedvaccinestrategy, knowing thepreciseIAV peptide

    targets recognized by

    T

    cell immunity

    after infection

    will

    be

    key. Lee and colleagues [5] utilized ex vivo stimulation of

    human

    peripheral

    blood

    mononuclear cells (PBMCs) with

    overlapping

    peptides

    that spanned the whole

    IAV protein

    spectrum, to show that individuals who had not been ex-

    posed

    to

    theH5N1

    virus

    (i.e., seronegativefor

    the

    virus)

    had

    both

    CD4+ and CD8+ T

    cells that could recognize peptides

    derived from this highly pathogenic virus. This suggested

    thatprevious

    infection

    by seasonal

    influenzacould generate

    T cell immunity that was capable of recognizing serological-

    lyunrelated IAV

    subtypes.

    Moreover, theyalsodemonstrat-

    ed that the major T cell targets were derived from the IAV

    Review

    1471-4906/

    2014 Elsevier Ltd. All rights reserved. http://dx.doi.org/10.1016/j.it.2014.06.004

    Corresponding author: Turner, S.J. ([email protected]).

    Keywords: influenza A virus; CD8+ T cell; CD4+ T cell; immunological memoryMHC

    class I MHC class II vaccination.

    396 Trends in Immunology, August 2014, Vol. 35, No. 8

    http://dx.doi.org/10.1016/j.it.2014.06.004mailto:[email protected]:[email protected]://dx.doi.org/10.1016/j.it.2014.06.004http://crossmark.crossref.org/dialog/?doi=10.1016/j.it.2014.06.004&domain=pdf
  • 8/11/2019 T Cell Mediated Immunity to Influenza

    2/7

    matrix protein (M)1 and nucleoprotein (NP). Studies that

    have

    alsoused

    theapproachof

    ex

    vivo

    stimulation

    of

    humanPBMCs withoverlapping IAV peptides have since confirmed

    thatpeptides

    fromtheM1

    andNP

    are themajor

    targetsfor

    T

    cell immunity [68]. Thus, pre-existing IAV-specific T cell

    immunity

    inducedbyinfectionwith one

    strainmay

    have the

    capacity (through

    cross-reactivity with conserved epitopes

    from a limited number ofviral proteins) to limit infection by

    different

    strains or

    subtypes, particularly in

    the absence

    of

    any

    neutralizing antibody

    responses.

    Moreover,novelT

    cell

    based vaccinestrategieswouldonlyneedto includea limited

    number

    of

    protein

    targets

    to

    ensure

    broad-based

    immunity

    and likelymakevaccineformulation a morestraightforward

    exercise than

    having

    to

    use

    something

    like whole

    inacti-

    vated IAV.In terms of precise

    peptide targets recognized by

    human

    memory

    T

    cells, there

    remains much

    to be

    learned. For

    example, a robust CD8+ T cell response against a peptide

    derived

    from the

    IAV

    matrix

    protein

    (M1,

    residues

    5866;

    M15866) can be readily detectedwithin HLA-A2+ individuals

    [9]. Given the repeateddemonstrationof this responseacross

    HLA-A2+ individuals,

    it

    is

    considered a

    dominant

    response.

    Are

    other

    suchdominant

    responses

    prevalent

    within

    individ-

    uals with other MHC haplotypes, and what is the full reper-

    toire of IAV

    peptides eliciting

    a

    CD8+ T

    cell

    response? Chen

    and

    colleagues

    addressed these questions by

    taking a

    sys-

    tematic approach

    wherein

    peripheral humanT

    lymphocytes

    fromseveralhealthydonors were cocultured with live IAV as

    an antigenic stimulus whereby infected

    PBMCs

    self pre-

    sented

    IAV antigens

    to pre-existing memory T

    cells. They

    then screened

    T

    cell

    reactivity against individual

    influenza

    proteins, enabling themtonarrowthe candidate pool, so that

    they could

    then define the

    minimal peptide

    targets after in

    vitro stimulation with overlapping peptides [10]. As previ-

    ouslyreported[11], the dominant CD8+T cell responses from

    different

    individuals targeted the

    conserved

    matrix

    (M) andnucleoprotein (NP) proteins. Importantly, using this system-

    atic approach,

    new

    peptide targets presented

    by an

    array

    of

    distinct

    HLA

    molecules

    were

    identified

    and

    found

    to be

    at

    times more prominent than the benchmark HLA-A2-M1

    epitope. Interestingly, it

    was noted

    that

    at

    least three

    of

    the newly identified IAV-peptide targets identified were

    longer than

    typical

    MHC class I binding peptides, and

    hence

    would not

    have been

    identified using

    established

    epitope

    prediction algorithms [10].

    These

    results

    suggest

    that a

    more

    systematic

    and direct

    approach,

    such

    as

    that outlined in

    the study

    by

    Chen and

    colleagues [10], is needed if peptide targets for a range of

    diverse

    MHC

    alleles

    are to

    be

    identified

    for

    possible inclu-

    sion in T cellbased vaccine approaches.Although the use ofspecificpeptides

    in

    vaccinesis

    a

    direct

    way

    of targetingT

    cell

    response,theapplication of targetedpeptide based vaccines

    will

    likely be

    limited by the fact that potential

    epitope

    targetswill

    be

    missed,

    as

    well

    as

    by

    thedifficulty

    of

    ensuring

    adequate coverage across numerous HLA subtypes. More-

    over,

    there

    is

    an

    increasing appreciation

    that IAV-specific

    T

    cell immunity

    may

    in

    fact drive

    immune

    escape

    in

    targeted

    T

    cell epitopes (Box 2). Alternatively, vaccine strategies that

    incorporate

    whole

    protein

    antigens, rather than peptides,

    would ensure

    adequate

    antigenic coverage across different

    HLA types. Aside from ensuring broad T cell immunity,

    another

    advantage

    of

    whole

    protein

    vaccinationagainst

    IAV

    would

    be

    thepotential

    to

    maximize

    fully

    humoral

    responseseither against conserved proteins,

    such

    as

    MP

    or

    NP

    [12],

    or

    conserved protein structures such as the stem region of the

    hemagglutinin

    (HA)

    [13];

    bothof

    which

    have

    shown

    promise

    in

    protection

    from heterologous

    challenge.

    It is important to note that whole proteinvaccine strat-

    egies do

    not

    circumvent

    the

    need

    for

    high-resolution

    epi-

    tope

    identification.

    A

    major

    driver

    is

    the

    increasing

    need

    to

    be able to track antigen-specific T cell responses after

    vaccination

    by

    use

    of

    soluble

    pMHC

    tetramer

    reagents

    [14]. The identification of new IAV-specific pMHC com-

    plexes, combined with recent advances in flow cytometric

    techniques that enable multiple specificities/parameters to

    be

    measured

    from

    a

    single

    blood

    sample

    [15,16], means

    we

    are

    potentially

    at

    the

    beginning

    of

    an

    era

    that

    will

    provide

    an unprecedented level of information about the kinetics,

    function

    and

    persistence

    of

    IAV-specific

    T

    cell

    immunity.

    Role of T cell immunity against IAV infection: lessons

    from humans

    Although

    IAV-specific

    CD4+ and

    CD8+ T

    cells

    are

    readily

    identifiable in humans [5,1720], their precise role in

    controlling

    IAV

    infection

    is

    unclear.

    A retrospective

    analy-

    sis

    demonstrated

    that

    prior

    symptomatic

    A(H1N1)

    infec-

    tion

    was

    associated

    with

    increased

    protection

    from

    the

    1957 A(H2N2) pandemic virus in adults but not children,

    suggesting

    an

    accumulation

    of

    heterologous

    immunity

    Box 1. Viral escape from cellular immunity: a paradox of

    acute

    infections

    DCs are a specialized subset of antigen-presenting cells that are key

    for alerting the host to infection and initiating T cell responses. DCs

    exist as twogeneral populations; those located in peripheral tissues

    and those located within secondary lymphoid tissues such as the

    lymph nodes. At least within the murine system, DCs located within

    these locations can be further divided into distinct subsets, with

    each reported to have distinct roles in antigen presentation andpriming of T cell responses [58]. DCs within the lymph nodes can be

    broadly separated into theCD11b+ CD8a or CD11b CD8a+ subsets,

    with the CD8a+ DCs being most efficient at presenting influenza

    antigens and activating nave, virus-specific T cells after infection

    [59]. Within peripheral t issues, DCs can be divided into CD103+

    CD11b or CD103 CD11b+ DCs, with the CD103+ DCs capable of

    migrating most efficiently to the draining lymph nodes after IAV

    infection [60]. Importantly, in the context of IAV infection, both lung-

    derived and lymph-node-derived DCs appear to play roles in the

    induction of T cell immunity to influenza [58,61]. For CD8+ T cell

    responses, both the CD8a+ CD11b (LN) and CD103+ CD11b (lung)

    DC subsets are important for priming [6062]. Although priming of

    CD8+ T cell responses is essentially limited to CD8a+ (LN) and/or

    CD103+ (lung) derived DCs, a broader range of DC subsets are

    capable of presenting antigen to CD4+ T cells [63]. However, in the

    context of IAV infection, the migratory CD11b

    CD103+

    DCs derivedfrom the lung are capable of activating nave CD4

    + T cell responses

    [62,64]. Although activated B cells can also present antigen to CD4+

    T cells, the key purpose of this TB interaction is the promotion of

    effective antibody responses, rather than initial priming of nave

    CD4+ T cell responses. Finally, although there is little information

    regarding the role of human DC subsets in priming IAV-specific T

    cell responses, subsets analogous to the mouse CD8a and CD103+

    DC subsets have been identified in humans and are most efficient at

    priming nave CD8+ T cell responses [65,66]. Thus, determining

    whether these same DC subsets are key players in the initiation of

    IAV-specific T cell responses in humans after infection/vaccination is

    of great interest and relevance to T cell based vaccine design.

    Review Trends in Immunology August 2014, Vol. 35, No. 8

    397

  • 8/11/2019 T Cell Mediated Immunity to Influenza

    3/7

    with age [21]. Although the mechanism is unknown, the

    fact

    that

    protection

    was

    mediated

    in

    the

    absence

    of

    any

    crossreactive

    antibody

    responses

    (because

    it

    was

    a

    pan-demic

    event),

    strongly

    suggests

    a

    key

    role

    for

    T

    cell

    medi-

    ated protection [21].

    The

    earliestindirectevidence

    in

    humans

    thatCD8+T

    cell

    immunity

    is

    important for

    protection against

    influenza-

    mediated illness came from a challenge study in which

    volunteerswere

    intranasally

    infected

    with

    a

    live, attenuat-

    ed

    IAV,

    and

    viral

    shedding

    was

    measured

    in

    clinicalsamples

    [20]. Decreased viral shedding was associated with a con-

    comitant increase in

    IAV-specific

    CD8+ T

    cell

    responses in

    volunteers who

    lacked neutralizing, strain-specific

    antibo-

    dies.

    These

    findings

    implied that IAV-specific

    CD8+ T

    cell

    responses could effectively limit primary IAV infection.

    The

    recent

    2009

    H1N1

    pandemic

    (2009

    pdmH1N1)

    provided

    a

    unique

    opportunity

    to

    determine

    whether

    pre-existing CD8+ T cell immunity provides protection

    from

    heterologous

    IAV

    infection.

    In

    one

    particular

    study

    [7],

    a

    cohort

    of

    individuals

    that

    lacked

    pre-existing

    anti-

    bodies to the 2009 H1N1 IAV pandemic were followed

    during

    pandemic

    cycles

    to

    determine

    whether

    pre-existing

    circulating

    memory

    T

    cell

    populations

    correlated

    with

    less

    severe disease outcomes after 2009 pdmH1N1 infection.

    Individuals who developed mild or no symptoms after 2009

    pdmH1N1 influenza infection were found to have higher

    circulating

    levels

    of

    pre-existing

    IAV-specific

    CD8+ effector

    memory T cells (defined by CD45RA+ CCR7 expression).

    Functionally,

    these

    effector

    memory

    CD8+ T

    cells

    exhibited

    the

    capacity

    to

    produce

    interferon

    (IFN)-g

    and

    were

    capa-

    ble

    of

    direct

    cytotoxicity

    against

    infected

    target

    cells.

    In-

    terestingly,

    there

    was

    no

    significant

    correlation

    between

    symptom severity (symptoms noted were runny nose, fe-

    ver,

    and

    sore

    throat)

    and

    the

    presence

    of

    pre-existing

    IAV-

    specific

    CD4+ T

    cells.

    Thus,

    in

    the

    setting

    of

    natural

    infec-

    tion, when antibody immunity is lacking, elevated influen-

    za-specific

    CD8

    +

    T

    cell

    immunity

    appears

    to

    help

    limit

    bothdisease symptoms and the spread of the virus.

    As

    noted

    earlier,

    both

    CD4+ and

    CD8+ T cell

    responses

    can

    target

    relatively

    conserved

    internal

    influenza

    proteins,

    implying that CD4+ T cells may have the potential to

    provide

    IAV-specific

    heterologous

    immunity

    [5]. To

    test

    this directly, Wilkinson and colleagues [8] challenged

    volunteers

    with

    either

    a

    wild

    type

    H1N1

    or

    a

    H3N2

    sea-

    sonal

    IAV

    strain,

    and

    then

    measured

    clinical

    symptoms

    and viral shedding over the course of infection. All volun-

    teers

    were

    seronegative

    for

    their

    respective

    challenge

    strain,

    therefore,

    the

    levels

    of

    pre-existing

    memory

    T

    cell

    responses were measured to determine their relation with

    disease

    progression.

    Although

    no

    significant

    correlation

    was found between symptom severity and presence ofpre-existing

    IAV-specific

    CD4+ T

    cells

    [7],

    Wilkinson

    et al. found a significant inverse correlation between dis-

    ease

    severity

    and

    pre-existing

    levels

    of

    circulating

    IAV-

    specific

    CD4+ cells.

    It

    is

    not

    clear

    why

    these

    two

    recently

    published IAV challenge studies came to different conclu-

    sions

    about

    the

    respective

    roles

    of IAV-specific

    CD8+ and

    CD4+ T

    cells.

    It

    might

    be

    the

    fact

    that

    Sridhar

    and

    collea-

    gues examined a natural experiment, whereas Wilkinson

    et al. analyzed heterologous IAV-specific T cell responses in

    the context

    of

    an

    experimental

    challenge

    of human

    volun-

    teers. Alternatively, analysis of T cell populations found

    within

    the

    respiratory

    tract

    during

    IAV

    infection

    may

    provide

    stronger

    correlates

    of

    immunity.

    Although

    morestudies

    like

    these

    are

    needed

    before

    any

    definitive

    answer,

    these findings nevertheless provide strong impetus for

    further

    developing

    an

    understanding

    of both

    CD8+ and

    CD4+ T

    cell

    effector

    functions

    and

    their

    role

    in

    IAV

    control.

    Mechanisms of CD8+ T cell dependent control of IAV

    infection

    Signaturevirus-specific CD8+T cell effectorfunctionsinclude

    the

    ability to

    produce

    a

    variety

    of cytotoxicmolecules

    such

    as

    perforin (Pfp) and granzymes (gzm), as well as being able to

    secrete a variety of potent inflammatory cytokines such as

    tumor necrosis factor (TNF)a and IFN-g (Figure 1). It is

    natural

    to expect that

    perhaps

    many,

    if

    not

    all,

    of

    these

    effector

    functions

    contribute to the

    limiting

    and

    eventual

    clearance of IAV infection. Pfp-deficient mice display an

    impaired

    capacity to clear

    IAV

    infection,

    suggesting

    that

    Pfp-dependentcytotoxicityplays

    a

    majorrole

    [22]. Unexpect-

    edly, mice deficient in the major granzyme proteins,A and B,

    do not

    show heightened susceptibility

    and

    can

    control IAV

    infection

    as

    effectively

    as

    wild

    type mice

    [23]. This

    suggests

    that other cell death pathways, such as FasFas ligand

    interactions

    mediated by

    activated T

    cells

    [22]

    or

    other

    death-domain-containing

    proteins

    such

    as

    TNF-related apo-

    ptosis inducing

    ligand

    (TRAIL) [24], may

    havea

    role. Amore

    intriguing possibility is that other granzymes, such as grzK,

    can compensate for

    the

    loss of grzA

    and

    B

    and

    contribute to

    Box 2. Viral escape from cellular immunity: a paradox of

    acute

    infections

    The acute nature of IAV infection is not typically considered to be a

    strong driver of mutational escape within targeted T cell epitopes

    because the duration of virus infection and the subsequent immune

    response is thought to be insufficient for the outgrowth of viral

    escape mutants. However, recent studies that examined the

    evolution of amino acid sequences within the relatively conserved

    NP from an array of different IAV isolates taken over the past 40years reported a high frequency of mutation. It was subsequently

    shown that these amino acid changes within known NP-derived

    CD8+ T cell epitopes resulted in loss of CD8+ T cell recognition of

    IAV-infected cells [67]. Importantly, amino acid variation is not

    limited to a single CD8+ T cell epitope, with variations identified

    within a number of other known NP-derived CD8+ T cell peptides

    that bind numerous different human MHC class molecules [6770].

    In a C57BL/6J mouse model of IAV infection, it was recently

    demonstrated that viral variants containing mutations at the MHC

    class I position 5 anchor residue of the DbNP366 epitope emerged

    coincidently with the peak of the DbNP366-specific CD8+ T cell

    response. When these viral variants were used to inoculate MHC-

    mismatched Balb/c mice (and were thereby relieved of T cell

    immune pressure) the variant IAVs reverted back to the wild type

    NP sequence. These data demonstrate that even a primary CD8+

    T cell response to IAV has the capacity to exert sufficient immunepressure to select escape variants. Although not as extensively

    studied, there is also some evidence that mutationwithinCD4+ T cell

    epitopes can result in escape from IAV-specific CD4+ T cell

    recognition. Given that T cell epitopes tend to be more conserved

    between different IAV strains than those recognized by antibody

    immunity, these data emphasize the need to

    select carefully

    potential antigens for inclusion in any future vaccine strategy, so

    that although a range of T cell reactivates are included, it might be

    necessary to exclude antigens that, from an evolutionary perspec-

    tive, appear to be targets of immune T cell selection.

    Review Trends in Immunology August 2014, Vol. 35, No. 8

    398

  • 8/11/2019 T Cell Mediated Immunity to Influenza

    4/7

    limiting

    and

    eventual

    control

    IAVinfection.

    grzK

    is

    expressed

    at high frequency by

    both

    mouse

    and

    human

    virus-specific

    CD8+ T cells [2528], and is also expressed at high levels in

    IAV-specific

    CD8+ T

    cells

    in grzA/B-deficient

    mice

    [23]. It is

    possible that

    perhaps

    grzK

    is

    the

    key cytolyticmolecule

    that

    mediates CD8+ T cell killing of virus-infected cells, and it

    remains to be

    determined

    whether

    grzK

    can

    compensate

    for

    the loss of

    grzA

    or

    B.

    IAV-specific CD8+ T cells can simultaneously produce a

    variety

    of

    proinflammatory

    cytokines

    in

    response

    to

    anti-

    gen activation

    [29]. Effector

    CD8+ T

    cells

    isolated

    from

    bronchoalveolar

    lavage

    exhibit

    a

    heightened

    functional

    capacity, particularly in terms of proinflammatory cyto-

    kine

    production

    [25,29]. Secretion

    of

    these

    mediators

    preferentially

    occurs

    at

    sites

    of

    active

    infection

    where

    there

    is

    increased

    presentation

    of

    viral

    determinants

    and

    a

    pre-

    existing inflammatory environment as a consequence of

    innate

    inflammatory

    mediators

    [30]. At

    least

    in

    mouse

    experimental

    systems,

    lung-resident

    memory

    T

    cells

    can

    persist long term after IAV infection, where they are

    thought

    to

    constitute

    a

    frontline

    defense

    against

    secondary

    challenge

    [31]. Following

    secondary

    IAV

    challenge,

    the

    lung-resident memory T cells are supplemented by chemo-

    kine

    CC

    receptor

    (CCR)5-dependent

    recruitment

    of

    circu-

    lating

    memory

    CD8+ T

    cell

    to

    the

    infected

    lung

    [32]. Both

    the

    resident

    memory

    and

    newly

    recruited

    IAV-specific

    CD8+ T cells can immediately secrete IFN-g upon antigen

    recognition

    and

    contribute

    to

    early

    virus

    elimination

    [33].

    (A)

    Lung airways

    Cytokine

    TH

    1 TH

    17

    Cytotoxic

    FasL

    Trail

    MHCI

    MHCII

    IL-10

    Perforin

    Gzm A, B K

    Perforin

    Gzm A, B

    IL-2

    IFN

    IFN IL-17A

    IL-6

    IL-21

    TNF

    TNF-

    IL1-

    CXCL9

    CCL2

    Lung parenchyma

    CCR5+CCR5+

    Blood vessels

    (B)

    (C)

    (D)

    TRENDS in Immunology

    Figure 1.

    T cell effector mechanisms in the IAV infected lung. (A) Recently activated IAV-specific CD8+ and CD4+ T cells are recruited to infected lung tissue in a CCR5-

    dependent manner.During a secondary response, recruitment of newly activated memory T cells supplements lung-resident memory T cells that remainedin the lungafter

    resolutionof a primary infection. (B) IAV-specificCD8+ T cells recognize IAV-infected lungepithelial cells presentingMHCclass I molecules presenting IAV-derivedpeptides.

    Upon T cell receptor recognition, effector CD8+ T cells contribute to viral control and elimination via a combination of mechanisms including: (i) delivery of cytotoxic

    moleucles such as perforin and granzymes; (ii) secretion of proinflammatory cytokines such as IFN-g and TNF-a; and (iii) expression of death domain receptors FasL and

    TRAIL that can initiatecell death afterbinding to their respective ligands.At later stages of infection, IAV-specificCD8+ T cells canalsoexpress IL-10 as a wayof helping limit

    T cell dependent immunopathology in the lung. (C) IAV-specific effector CD4+ T cells contribute to viral control and elimination via secretion of either TH1 or TH17

    proinflammatory cytokines. UpregulationofMHCII on inflammed epithelial cells means thatCD4+ T cells candirectly recognize infected cells,and there is a

    suggestionthat

    lungeffectorCD4+ T cells mayalsomediatedirect cellcytotoxicity viadeliveryof perforinand granzymes. (D)ActivatedeffectorCD4+ T cells canalso trigger thesecretion of

    innate cytokines such as IL1-b, CXCL9, and CCL2 helping contribute to the proinflammatory response in the infected lung. The cellular source of these cytokines is not

    known are likely to be lung residentmacrophages. Abbreviations: CCL2, chemokine CC ligand 2; CCR5, chemokine CC receptor 5; CXCL9, chemokine CXC ligand 9; Gzm,

    granzyme; IAV, influenza A virus; FasL, Fas ligand; IFN, interferon; IL, interleukin; TH, T helper; TNF, tumor necrosis factor; TRAIL, TNF-related apoptosis inducing ligand.

    Review Trends in Immunology August 2014, Vol. 35, No. 8

    399

  • 8/11/2019 T Cell Mediated Immunity to Influenza

    5/7

    The

    recruitment

    to

    the

    murine

    lung

    of

    these

    highly

    active

    memory

    CD8+ T

    cells

    expressing

    IFN-g

    has

    been

    shown

    to

    be

    key

    for

    protection

    against

    influenza

    infection

    [34].

    The enhanced effector potential of lung localized CD8+T

    cell

    effectors

    also

    increases

    the

    risk

    of

    damage

    to

    lung

    tissue

    due

    to

    excessive

    inflammation

    (reviewed

    in [35]).

    Thus, to mitigate damage to the sensitive lung tissue by

    potent

    T

    cell

    effector

    functions,

    a

    balance

    must

    be

    struckbetween ensuring effective antiviral potency while not

    causing

    immunopathology.

    It

    is

    intriguing

    that

    IAV-spe-

    cific

    CD8+ effector

    T

    cells

    isolated

    from

    infected

    mouse

    lungs are capable of producing interleukin (IL)-10; a potent

    negative

    regulator

    of

    inflammation

    [36]. The

    ability

    of

    effector CD8+ T cells to produce IL-10 is dependent on

    migration

    into

    the

    inflamed

    lung

    [37], suggesting

    that

    there

    are

    signals

    specific

    to

    the

    infected

    lung

    microenvi-

    ronment that trigger regulatory functions in otherwise

    proinflammatory

    IAV-specific

    CD8+ T

    cells.

    In

    this

    way,

    the immune

    response

    can

    balance

    the

    need

    for

    inflamma-

    tion required to clear IAV infection, with the need to limit

    tissue

    injury

    by

    the

    inflammatory

    response.

    Mechanisms

    of

    CD4+ T

    cell

    control

    of

    viral

    infection

    Classically, activated CD4+ T cells are considered to be key

    for

    promotion

    of

    effective

    antibody

    responses

    via

    support

    of

    germinal

    center

    formation

    that

    results

    in

    affinity

    matura-

    tion and isotype switching [3841]. This occurs through the

    provision

    of

    key

    co-stimulatory

    signals

    such

    as

    inducible

    T

    cell

    co-stimulator

    (ICOS),

    and

    the

    production

    of

    cytokines

    such as IL-21 [40,41]. The antibody response to IAV infec-

    tion

    is

    critical

    for

    protection

    [42]; lack

    of

    neutralizing

    antibody

    levels

    in

    the

    population

    is

    a

    key

    factor

    controlling

    the emergence of IAV pandemics [43].

    Thefinding

    that

    memory

    CD4+ T

    cell

    responses

    contrib-

    ute

    to

    heterosubtypic

    immunity

    against

    a

    potential

    pan-demic

    IAV

    [8]

    suggests

    that

    memory

    CD4+ T

    cells

    play

    a

    key role in the control of IAV infection, but the mechanisms

    involved

    are

    not

    clear.

    Adoptive

    transfer

    of

    a

    large

    number

    ofex vivo isolated memory IAV-specific CD4+ T cells into a

    mouse model of infection augmented both IAV-specific

    CD8+ and

    B

    cell

    responses

    against

    primary

    infection

    [44]. A

    more

    detailed

    analysis

    of

    how

    memory

    CD4+ T

    cells

    can promote primary IAV-specific B cell response has

    shown

    that

    establishment

    of

    NP-specific

    memory

    CD4+

    T cells by peptide vaccination of mice promoted robust

    germinal center formation and a more rapid primary

    NP-specific antibody response after IAV infection, com-

    pared

    to

    unvaccinated

    mice

    [45]. Strikingly,

    memory

    NP-specific

    CD4+ T

    cells

    did

    not

    promote

    antibody

    responses to otherviral proteins, including the HA protein,

    the

    major

    target

    of

    the

    antibody

    response.

    This

    suggests

    that

    both

    the

    antibody

    and

    T

    cell

    responses

    are

    linked

    to

    the sameviral target; likely as a consequence of the ability

    of B

    cells

    to

    process

    and

    present

    CD4+ T

    cell

    epitopes

    from

    antigen

    captured

    and

    internalized

    via

    surface

    immuno-

    globulin receptors.

    Mouse

    models

    of

    IAV

    infection

    provide

    a

    tool

    whereby

    CD4+ T

    cell

    effector

    mechanisms

    can

    be

    delineated

    more

    precisely

    (reviewed

    in [46]). For

    example,

    recent

    studies

    have utilized adoptive transfer of T cell receptor (TCR)

    transgenic

    CD4+ T

    cells

    specific

    for

    an

    epitope

    of

    the

    HA

    protein

    of

    an

    H1N1

    IAV [A/PR8/34

    (HNT)]

    to

    determine

    the contribution

    of

    CD4+ T

    cells

    to

    protection

    from

    IAV

    infection

    [44].

    Initial experiments

    demonstrated that

    adoptive transfer of CD4+ HNT T cells that were differen-

    tiated in vitro into proinflammatory T helper (TH)1 or

    TH17 lineages (Figure 1) were more capable of mediating

    clearance and protection from IAV infection, compared to

    uncommitted

    (TH

    0) or

    anti-inflammatory (TH

    2) CD4

    +

    effectors [44]. This control was partly via augmentation

    of endogenous IAV-specific

    CD8+ T

    cell

    and B cell

    responses

    and

    partly

    via triggering expression

    of innate

    cytokines such as IL-1b, IL-6, chemokine CXC ligand

    (CXCL)9 and chemokine CC

    ligand (CCL)2, particularly

    within the infected lung[47].

    The

    demonstration

    that

    at

    least

    in

    murine

    models,

    influenza

    infection

    can

    induce in vivo MHC class II expres-

    sion on lung epithelial cells [48] highlights the possibility

    that

    CD4+ T

    cells

    could

    have

    a

    role

    in

    control

    of

    IAV

    infection

    by

    directly

    recognizing

    and

    eliminating

    virus-

    infected targets. In fact, protection from IAV infection

    conferred

    by

    adoptive

    transfer

    of

    memory

    HNT

    CD4+ T

    cells into immunodeficient mice was abrogated when thesecells

    were

    deficient

    in

    either

    IFN-g

    or

    perforin

    [44]. Hence,

    aside from providing help to B cells and CD8+ T cells, IAV-

    specific

    CD4+ T

    cells

    have

    the

    capacity

    to

    target

    directly

    IAV-infected

    cells,

    thereby

    contributing

    to

    the

    control

    and

    elimination of IAV infection. Such unconventional mecha-

    nisms

    of

    T

    cell

    action

    must

    be

    appreciated

    for

    the

    strategic

    design

    of

    vaccines

    aiming

    to

    elicit

    effective

    cellular

    immu-

    nity.

    CD4+ T cell regulation of IAV-specific CD8+ T cell

    responses

    The precise

    role

    of

    CD4+ T cells

    in

    promoting

    and

    regulat-

    ing

    CD8+

    T

    cell

    responses

    induced

    by

    IAV

    infection

    was,until

    recently,

    enigmatic.

    This

    is

    partly

    because

    inmice,

    an

    effective primary CD8+ T cell response to IAV can be

    induced

    independently

    of

    CD4+ T

    cells

    [49]. In

    this

    case,

    direct

    activation

    of

    dendritic

    cells

    (DCs)

    via

    the

    engage-

    ment of Toll-like receptors (TLRs) by IAV circumvents the

    need

    for

    CD4+ TH-dependent CD40 ligand (CD40L) licens-

    ing of DCs

    to

    promote

    primary

    virus-specific

    CD8+ T

    cell

    responses [50].

    However, memory

    CD8+ T

    cells

    that

    are

    primed

    in

    the

    absence of CD4+T cells are reduced in number and show an

    inability to response to secondary infection, compared to

    memory CD8+ T cells primed in the presence of CD4+ T

    cells

    [49]. So,

    although

    dispensable

    for

    primary

    activation

    and expansion,

    CD4+ T

    cell

    help

    is

    crucial

    (during

    the

    initial priming phase) for programming optimal IAV-spe-

    cific

    CD8+ T

    cell

    memory.

    The

    role

    of

    CD4+ T

    cell

    help

    in

    this

    case

    is

    the

    provision

    of

    co-stimulatory

    signals

    via

    CD40LCD40-dependent interactions with DCs that lead

    to optimal

    priming

    of

    the

    IAV-specific

    CD8+ T

    cell

    re-

    sponse.

    These

    signals

    received

    from

    licensed

    DCs

    then

    ensure the responding CD8+ T cells are capable of auto-

    crine

    IL-2

    production

    [51], which

    is

    crucial

    for

    their

    surviv-

    al into

    memory.

    What

    remains

    unclear

    are

    the

    precise

    signals

    provided

    by

    CD4-dependent

    licensing

    of

    DCs

    that

    ensure responding CD8+ T cells can establish effective

    memory

    populations.

    Uncovering

    these

    mechanisms

    would

    Review Trends in Immunology August 2014, Vol. 35, No. 8

    400

  • 8/11/2019 T Cell Mediated Immunity to Influenza

    6/7

    provide

    information

    crucial

    to

    the

    design

    of

    any

    CD8+ T

    cell

    based

    vaccine

    strategy

    to

    promote

    optimal

    memory

    formation.

    CD4+ T regulatory (Treg) cells have been shown to limit

    effector

    CD8+ T

    cell

    differentiation

    in

    response

    to

    virus

    infection

    and

    immunization

    [5254],

    and

    they

    have

    the

    capacity to suppress potently primary IAV-specific CD8+ T

    cell

    responses

    after

    infection

    of

    mice

    [55,56]. So,

    how

    is

    aneffective primary CD8+ T response sustained in the face of

    CD4+ Treg

    cell

    mediated

    suppression?

    Recent

    work

    from

    Randall

    and

    colleagues

    suggests

    that

    activation

    of

    CD40L+

    CD4+ THcells early after IAV infection is key to ensuring

    appropriate

    DC

    activation

    that

    serves

    to

    limit

    the

    expan-

    sion and activation of Treg cells [55], which in turn limits

    Treg

    cell

    suppression

    of

    the

    primary

    CD8+ T

    cell

    response

    during

    the

    early

    phases

    of

    infection.

    As

    the

    infection

    is

    cleared and antigen becomes limiting, Treg cells begin to

    exert

    their

    suppressive

    effects

    and

    effectively

    promote

    the

    tapering

    of

    the

    effector

    CD8+ T

    cell

    response

    during

    the

    contraction phase [55]. In this way, Treg cells may limit

    potential

    damage

    caused

    by

    a

    prolonged

    CD8+ T

    cell

    re-

    sponse at later stages of infection. This mechanism is onlyrecently

    described

    and

    raises

    several

    questions.

    For

    exam-

    ple, is the induction of Treg cells diminished in the case of

    highly

    pathogenic

    IAV

    infection,

    where

    increased

    immu-

    nopathology

    is

    associated

    with

    highly

    pathogenic

    H5N1

    or

    the recent H7N9 infection of humans?

    The

    overall

    picture

    is

    that

    is

    that

    all

    arms

    of

    the

    adaptive

    response

    have

    a

    role

    to

    play

    in

    the

    control

    of

    IAV infection. B cell/antibody-mediated immunity plays

    the

    major

    role

    when

    it

    comes

    to

    preventing

    infection

    with

    antigenically

    matched

    strains.

    In

    cases

    where

    antibody

    reactivity is limiting or absent, there is mounting evidence

    that

    both

    CD8+ and

    CD4+ T

    cells

    have

    key

    roles

    in

    limiting

    IAV

    infection,

    particularly

    in

    the

    case

    of

    heterologous

    IAVchallenge.

    What

    has

    begun

    to

    emerge,

    yet

    remains

    incom-

    pletely understood, is the range and redundancy of mech-

    anisms

    utilized

    by T

    cells

    in

    both

    controlling

    infection

    and

    limiting

    immunopathology,

    as

    well

    as

    the

    precise

    interac-

    tions between the adaptive immune cell populations. When

    considering

    the

    development

    of

    new

    vaccines

    for

    engaging

    heterologous

    T

    cell

    immunity,

    it

    is

    essential

    that

    these

    features of T cell activity be understood to ensure estab-

    lishment

    of

    an

    effective

    memory

    T

    cell

    population.

    Concluding remarks

    Although there has long been acknowledgement that cel-

    lular

    immunity

    to

    IAV

    plays

    a

    role

    in

    protection

    from

    infection,

    it

    is

    only

    with

    recent

    advances

    in

    the

    identifica-

    tion and isolation of IAV-specific T cells that this has been

    accepted

    as

    an

    important

    immunological

    correlate

    of

    pro-

    tection

    from

    IAV

    infection.

    Given

    the

    extremely

    high

    mu-

    tation rate of the influenza proteins (NA and HA) typically

    targeted

    by

    antibodies,

    it

    is

    becoming

    clear

    that

    protection

    from

    IAV

    infection,

    and

    a

    broader

    range

    of

    infections

    such

    as HIV, hepatitis Cvirus, and malaria, will requirevaccine

    strategies

    that

    induce

    robust

    and

    long-lived

    T

    cell

    responses

    [57]. The

    improved

    capacity

    to

    enumerate

    and

    isolate

    IAV-specific

    T

    cell

    responses

    has

    also

    allowed

    great-

    er insight into the dynamics, location, gene expression, and

    genomic

    organization

    of

    IAV-specific

    T

    cell

    immunity

    at

    distinct

    stages

    of

    T

    cell

    immune

    response

    [57]. Although

    such

    analyses

    are

    greatly

    enhancing

    our

    understanding

    of

    both

    molecular

    regulation

    and

    immune

    mechanisms,

    the

    practical challenge of how best to manipulate both CD4+

    and

    CD8+ T

    cell

    responses,

    particularly

    via

    vaccination,

    to

    achieve

    a

    measure

    of

    long-term,

    if

    partial,

    heterosubtypic

    protection is still ahead of us.

    Acknowledgments

    This work is supported by an Australian Research Council Future

    Fellowship (awarded to S.J.T.); a Sylvia and Charles Viertal Senior

    ResearchFellowship (awarded to N.L.L.); Australian NationalHealth and

    Medical Research Council (NHMRC) program grant 5671222 (awarded to

    S.J.T.) and NHMRC project grant AI1046333 (awarded to N.L.L.).

    References1 Doherty, P.C. and Christensen, J.P. (2000) Accessing complexity: the

    dynamics of virus-specific T cell responses. Annu. Rev. Immunol. 18,

    561592

    2 Strutt, T.M. et al. (2013) Multipronged CD4+ T-cell effector and

    memory responses cooperate to provide potent immunity against

    respiratory virus. Immunol. Rev. 255, 149164

    3 Thomas, P.G. et al. (2006) Cell-mediated protection in influenzainfection. Emerg. Infect. Dis. 12, 4854

    4 Doherty, P.C.et al. (2006)Influenza andthe challenge forimmunology.

    Nat. Immunol. 7, 449455

    5 Lee, L.Y. et al. (2008) Memory T cells established by seasonal human

    influenza A infection cross-react with avian influenza A (H5N1) in

    healthy individuals. J. Clin. Invest. 118, 34783490

    6 Grant, E. et al. (2013) Nucleoprotein of influenza A virus is a major

    target of immunodominant CD8+ T-cell responses.Immunol. Cell Biol.

    91, 184194

    7

    Sridhar, S. et al. (2013) Cellular immune correlates of protection

    against symptomatic pandemic influenza. Nat. Med. 19, 13051312

    8 Wilkinson, T.M.et al. (2012) Preexistinginfluenza-specificCD4+T cells

    correlate with disease protection against influenza challenge in

    humans. Nat. Med. 18, 274280

    9 Gotch, F.et al. (1987)Cytotoxic T lymphocytes recognize a fragment of

    influenza virusmatrixprotein in associationwith HLA-A2.Nature 326,

    881882

    10 Wu, C. et al. (2011) Systematic identification of immunodominant

    CD8+ T-cell responses to influenza A virus in HLA-A2 individuals.

    Proc. Natl. Acad. Sci. U.S.A. 108, 91789183

    11 Nayak, J.L.et al. (2010)Analysesof thespecificity ofCD4T cells during

    the primary immune response to influenza virus reveals dramatic

    MHC-linked asymmetries in reactivity to individual viral proteins.

    Viral Immunol. 23, 169180

    12 Alam,S.et al. (2014)CD4T cell help is limitingandselective duringthe

    primary B cell response to influenza virus infection.J. Virol. 88, 314

    324

    13 Corti, D. et al. (2010) Heterosubtypic neutralizing antibodies are

    produced by individuals immunized with a seasonal influenza

    vaccine. J. Clin. Invest. 120, 16631673

    14 Guillaume, P. et al. (2009) Soluble MHC-peptide complexes: tools for

    themonitoring of T cell responses in clinical trials and basic research.

    Cancer Immun. 9, 7

    15 Hadrup, S.R. et al. (2009) Parallel detection of antigen-specific T-cell

    responses by multidimensional encoding of MHC multimers. Nat.

    Methods 6, 520526

    16 Newell, E.W. et al. (2012) Cytometry by time-of-flight shows

    combinatorial cytokine expression and virus-specific cell niches

    within a continuum of CD8+ T cell phenotypes.Immunity 36, 142152

    17 Forrest, B.D. et al. (2008) Correlation of cellular immune responses

    with protection against culture-confirmed influenza virus in young

    children. Clin. Vaccine Immunol. 15, 10421053

    18 Kreijtz, J.H.et al. (2007) Primary influenza A virus infection induces

    cross-protective immunity against a lethal infection with a

    heterosubtypic virus strain in mice. Vaccine 25, 612620

    19 Kreijtz, J.H. et al. (2008) Cross-recognition of avian H5N1 influenza

    virus by human cytotoxic T-lymphocyte populations directed to human

    influenza A virus. J. Virol. 82, 51615166

    Review Trends in Immunology August 2014, Vol. 35, No. 8

    401

    http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0005http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0005http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0005http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0005http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0005http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0010http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0010http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0010http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0010http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0010http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0010http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0010http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0015http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0015http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0015http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0015http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0015http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0015http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0020http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0020http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0020http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0020http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0020http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0025http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0025http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0025http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0025http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0025http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0025http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0025http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0030http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0030http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0030http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0030http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0030http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0030http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0030http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0035http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0035http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0035http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0035http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0035http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0035http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0040http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0040http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0040http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0040http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0040http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0040http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0045http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0045http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0045http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0045http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0045http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0045http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0045http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0050http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0050http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0050http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0050http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0050http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0050http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0050http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0055http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0055http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0055http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0055http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0055http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0055http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0055http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0060http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0060http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0060http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0060http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0060http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0060http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0060http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0065http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0065http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0065http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0065http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0065http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0065http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0065http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0070http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0070http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0070http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0070http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0070http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0070http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0075http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0075http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0075http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0075http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0075http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0075http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0075http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0080http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0080http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0080http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0080http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0080http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0080http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0080http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0080http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0085http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0085http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0085http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0085http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0085http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0085http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0085http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0090http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0090http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0090http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0090http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0090http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0090http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0090http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0095http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0095http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0095http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0095http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0095http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0095http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0095http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0095http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0095http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0095http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0090http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0090http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0090http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0085http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0085http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0085http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0080http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0080http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0080http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0080http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0075http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0075http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0075http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0070http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0070http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0070http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0065http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0065http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0065http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0060http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0060http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0060http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0055http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0055http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0055http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0055http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0050http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0050http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0050http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0050http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0045http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0045http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0045http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0040http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0040http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0040http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0040http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0035http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0035http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0030http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0030http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0030http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0030http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0025http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0025http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0025http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0020http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0020http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0015http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0015http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0010http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0010http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0010http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0010http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0005http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0005http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0005
  • 8/11/2019 T Cell Mediated Immunity to Influenza

    7/7

    http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0350http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0350http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0350http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0350http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0345http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0345http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0345http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0345http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0340http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0340http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0340http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0335http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0335http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0335http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0335http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0330http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0330http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0330http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0330http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0330http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0325http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0325http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0325http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0325http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0325http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0320http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0320http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0320http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0320http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0320http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0315http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0315http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0310http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0310http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0310http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0310http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0305http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0305http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0305http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0305http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0300http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0300http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0300http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0300http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0295http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0295http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0295http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0295http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0290http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0290http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0290http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0285http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0280http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0280http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0280http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0280http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0275http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0275http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0275http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0275http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0275http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0270http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0270http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0270http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0270http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0270http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0265http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0265http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0265http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0265http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0265http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0265http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0260http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0260http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0260http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0260http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0260http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0255http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0255http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0255http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0250http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0250http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0250http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0245http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0245http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0245http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0245http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0240http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0240http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0240http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0235http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0235http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0235http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0230http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0230http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0230http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0225http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0225http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0225http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0225http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0220http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0220http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0220http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0220http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0215http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0215http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0215http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0210http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0210http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0205http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0205http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0205http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0200http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0200http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0200http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0195http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0195http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0195http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0190http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0190http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0190http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0185http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0185http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0185http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0180http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0180http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0180http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0175http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0175http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0175http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0170http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0170http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0170http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0170http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0165http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0165http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0165http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0165http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0160http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0160http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0160http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0160http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0155http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0155http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0150http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0150http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0145http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0145http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0145http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0145http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0145http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0140http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0140http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0140http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0135http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0135http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0135http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0130http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0130http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0130http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0130http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0125http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0125http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0125http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0120http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0120http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0115http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0115http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0115http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0110http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0110http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0110http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0105http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0105http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0105http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0100http://refhub.elsevier.com/S1471-4906(14)00109-4/sbref0100